Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Cell Sci ; 128(18): 3456-65, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26240177

RESUMEN

PKB/Akt activation is a common step in tumour growth, proliferation and survival. Akt activation is understood to occur at the plasma membrane of cells in response to growth factor stimulation and local production of the phosphoinositide lipid phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P3] following phosphoinositide 3-kinase (PI3K) activation. The metabolism and turnover of phosphoinositides is complex--they act as signalling molecules as well as structural components of biological membranes. The localisation and significance of internal pools of PtdIns(3,4,5)P3 has long been speculated upon. By using transfected and recombinant protein probes for PtdIns(3,4,5)P3, we show that PtdIns(3,4,5)P3 is enriched in the nuclear envelope and early endosomes. By exploiting an inducible dimerisation device to recruit Akt to these compartments, we demonstrate that Akt can be locally activated in a PtdIns(3,4,5)P3-dependent manner and has the potential to phosphorylate compartmentally localised downstream substrates. This could be an important mechanism to regulate Akt isoform substrate specificity or influence the timing and duration of PI3K pathway signalling. Defects in phosphoinositide metabolism and localisation are known to contribute to cancer, suggesting that interactions at subcellular compartments might be worthwhile targets for therapeutic intervention.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Membrana Celular/metabolismo , Activación Enzimática , Células HeLa , Humanos , Fosforilación , Transporte de Proteínas , Transducción de Señal , Especificidad por Sustrato , Transfección
2.
Biochem Soc Trans ; 42(5): 1435-40, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25233428

RESUMEN

Phosphoinositide-dependent kinase 1 (PDK1) is the master regulator of at least 23 other AGC kinases whose downstream signalling has often been implicated in various diseases and in particular in cancer. Therefore there has been great interest in determining how PDK1 is controlled and how it regulates its substrates spatially and temporally. The understanding of these mechanisms could offer new possibilities for therapeutic intervention. Over the years, a more comprehensive view of the mechanisms involved in the regulation of PDK1 has emerged and these comprise serine/threonine as well as tyrosine phosphorylation, subcellular localization, regulator binding and conformation status. In the present review, we discuss how various molecular mechanisms are together responsible for the conformational regulation behind the activation of PDK1 in cells.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Modelos Moleculares , Transducción de Señal , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/química , Animales , Dimerización , Activación Enzimática , Humanos , Ligandos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Fosforilación , Conformación Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Serina/metabolismo , Treonina/metabolismo , Tirosina/metabolismo
3.
Biochemistry ; 52(28): 4820-9, 2013 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-23745598

RESUMEN

Phosphoinositide-dependent kinase-1 (PDK1) is an essential master kinase recruited to the plasma membrane by the binding of its C-terminal PH domain to the signaling lipid phosphatidylinositol-3,4,5-trisphosphate (PIP3). Membrane binding leads to PDK1 phospho-activation, but despite the central role of PDK1 in signaling and cancer biology, this activation mechanism remains poorly understood. PDK1 has been shown to exist as a dimer in cells, and one crystal structure of its isolated PH domain exhibits a putative dimer interface. It has been proposed that phosphorylation of PH domain residue T513 (or the phospho-mimetic T513E mutation) may regulate a novel PH domain dimer-monomer equilibrium, thereby converting an inactive PDK1 dimer to an active monomer. However, the oligomeric states of the PH domain on the membrane have not yet been determined, nor whether a negative charge at position 513 is sufficient to regulate its oligomeric state. This study investigates the binding of purified wild-type (WT) and T513E PDK1 PH domains to lipid bilayers containing the PIP3 target lipid, using both single-molecule and ensemble measurements. Single-molecule analysis of the brightness of the fluorescent PH domain shows that the PIP3-bound WT PH domain on membranes is predominantly dimeric while the PIP3-bound T513E PH domain is monomeric, demonstrating that negative charge at the T513 position is sufficient to dissociate the PH domain dimer and is thus likely to play a central role in PDK1 monomerization and activation. Single-molecule analysis of two-dimensional (2D) diffusion of PH domain-PIP3 complexes reveals that the dimeric WT PH domain diffuses at the same rate as a single lipid molecule, indicating that only one of its two PIP3 binding sites is occupied and there is little penetration of the protein into the bilayer as observed for other PH domains. The 2D diffusion of T513E PH domain is slower, suggesting the negative charge disrupts local structure in a way that allows deeper insertion of the protein into the viscous bilayer, thereby increasing the diffusional friction. Ensemble measurements of PH domain affinity for PIP3 on plasma membrane-like bilayers reveal that the dimeric WT PH domain possesses a one order of magnitude higher target membrane affinity than the previously characterized monomeric PH domains, consistent with a dimerization-triggered, allosterically enhanced affinity for one PIP3 molecule (a much larger affinity enhancement would be expected for dimerization-triggered binding to two PIP3 molecules). The monomeric T513E PDK1 PH domain, like other monomeric PH domains, exhibits a PIP3 affinity and bound state lifetime that are each 1 order of magnitude lower than those of the dimeric WT PH domain, which is predicted to facilitate release of activated, monomeric PDK1 to the cytoplasm. Overall, the study yields the first molecular picture of PH domain regulation via electrostatic control of dimer-monomer conversion.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/química , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Dimerización , Activación Enzimática , Transferencia Resonante de Energía de Fluorescencia , Humanos , Membrana Dobles de Lípidos , Fosforilación , Estructura Terciaria de Proteína
4.
J Clin Oncol ; 41(14): 2561-2570, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-36821809

RESUMEN

PURPOSE: In many cancers, the expression of immunomodulatory ligands leads to immunoevasion, as exemplified by the interaction of PD-L1 with PD-1 on tumor-infiltrating lymphocytes. Profound advances in cancer treatments have come with the advent of immunotherapies directed at blocking these immuno-suppressive ligand-receptor interactions. However, although there has been success in the use of these immune checkpoint interventions, correct patient stratification for these therapies has been challenging. MATERIALS AND METHODS: To address this issue of patient stratification, we have quantified the intercellular PD-1/PD-L1 interaction in formalin-fixed paraffin-embedded tumor samples from patients with non-small cell lung carcinoma, using a high-throughput automated quantitative imaging platform (quantitative functional proteomics [QF-Pro]). RESULTS: The multisite blinded analysis across a cohort of 188 immune checkpoint inhibitor-treated patients demonstrated the intra- and intertumoral heterogeneity of PD-1/PD-L1 immune checkpoint engagement and notably showed no correlation between the extent of PD-1/PD-L1 interaction and PD-L1 expression. Importantly, PD-L1 expression scores used clinically to stratify patients correlated poorly with overall survival; by contrast, patients showing a high PD-1/PD-L1 interaction had significantly better responses to anti-PD-1/PD-L1 treatments, as evidenced by increased overall survival. This relationship was particularly strong in the setting of first-line treatments. CONCLUSION: The functional readout of PD-1/PD-L1 interaction as a predictive biomarker for the stratification of patients with non-small-cell lung carcinoma, combined with PD-L1 expression, should significantly improve the response rates to immunotherapy. This would both capture patients excluded from checkpoint immunotherapy (high PD-1/PD-L1 interaction but low PD-L1 expression, 24% of patients) and additionally avoid treating patients who despite their high PD-L1 expression do not respond and suffer from side effects.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Inmunoterapia/métodos , Antígeno B7-H1
5.
PLoS Biol ; 7(1): e17, 2009 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-19166270

RESUMEN

Protein kinase B (PKB/Akt) belongs to the AGC superfamily of related serine/threonine protein kinases. It is a key regulator downstream of various growth factors and hormones and is involved in malignant transformation and chemo-resistance. Full-length PKB protein has not been crystallised, thus studying the molecular mechanisms that are involved in its regulation in relation to its structure have not been simple. Recently, the dynamics between the inactive and active conformer at the molecular level have been described. The maintenance of PKB's inactive state via the interaction of the PH and kinase domains prevents its activation loop to be phosphorylated by its upstream activator, phosphoinositide-dependent protein kinase-1 (PDK1). By using a multidisciplinary approach including molecular modelling, classical biochemical assays, and Förster resonance energy transfer (FRET)/two-photon fluorescence lifetime imaging microscopy (FLIM), a detailed model depicting the interaction between the different domains of PKB in its inactive conformation was demonstrated. These findings in turn clarified the molecular mechanism of PKB inhibition by AKT inhibitor VIII (a specific allosteric inhibitor) and illustrated at the molecular level its selectivity towards different PKB isoforms. Furthermore, these findings allude to the possible function of the C-terminus in sustaining the inactive conformer of PKB. This study presents essential insights into the quaternary structure of PKB in its inactive conformation. An understanding of PKB structure in relation to its function is critical for elucidating its mode of activation and discovering how to modulate its activity. The molecular mechanism of inhibition of PKB activation by the specific drug AKT inhibitor VIII has critical implications for determining the mechanism of inhibition of other allosteric inhibitors and for opening up opportunities for the design of new generations of modulator drugs.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Regulación Alostérica/fisiología , Animales , Sitios de Unión/efectos de los fármacos , Regulación de la Expresión Génica , Ratones , Mutagénesis , Fosforilación , Conformación Proteica/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Relación Estructura-Actividad
6.
Nat Rev Cancer ; 21(1): 51-63, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33177705

RESUMEN

The maturing mutational landscape of cancer genomes, the development and application of clinical interventions and evolving insights into tumour-associated functions reveal unexpected features of the protein kinase C (PKC) family of serine/threonine protein kinases. These advances include recent work showing gain or loss-of-function mutations relating to driver or bystander roles, how conformational constraints and plasticity impact this class of proteins and how emergent cancer-associated properties may offer opportunities for intervention. The profound impact of the tumour microenvironment, reflected in the efficacy of immune checkpoint interventions, further prompts to incorporate PKC family actions and interventions in this ecosystem, informed by insights into the control of stromal and immune cell functions. Drugging PKC isoforms has offered much promise, but when and how is not obvious.


Asunto(s)
Neoplasias/enzimología , Proteína Quinasa C/fisiología , Animales , Humanos , Isoenzimas/fisiología , Mutación , Fosforilación , Regiones Promotoras Genéticas , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Microambiente Tumoral
7.
PLoS Biol ; 5(4): e95, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17407381

RESUMEN

Protein kinase B (PKB/Akt) is a pivotal regulator of diverse metabolic, phenotypic, and antiapoptotic cellular controls and has been shown to be a key player in cancer progression. Here, using fluorescent reporters, we shown in cells that, contrary to in vitro analyses, 3-phosphoinositide-dependent protein kinase 1 (PDK1) is complexed to its substrate, PKB. The use of Förster resonance energy transfer detected by both frequency domain and two-photon time domain fluorescence lifetime imaging microscopy has lead to novel in vivo findings. The preactivation complex of PKB and PDK1 is maintained in an inactive state through a PKB intramolecular interaction between its pleckstrin homology (PH) and kinase domains, in a "PH-in" conformer. This domain-domain interaction prevents the PKB activation loop from being phosphorylated by PDK1. The interactive regions for this intramolecular PKB interaction were predicted through molecular modeling and tested through mutagenesis, supporting the derived model. Physiologically, agonist-induced phosphorylation of PKB by PDK1 occurs coincident to plasma membrane recruitment, and we further shown here that this process is associated with a conformational change in PKB at the membrane, producing a "PH-out" conformer and enabling PDK1 access the activation loop. The active, phosphorylated, "PH-out" conformer can dissociate from the membrane and retain this conformation to phosphorylate substrates distal to the membrane. These in vivo studies provide a new model for the mechanism of activation of PKB. This study takes a crucial widely studied regulator (physiology and pathology) and addresses the fundamental question of the dynamic in vivo behaviour of PKB with a detailed molecular mechanism. This has important implications not only in extending our understanding of this oncogenic protein kinase but also in opening up distinct opportunities for therapeutic intervention.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Activación Enzimática , Transferencia Resonante de Energía de Fluorescencia , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Células 3T3 NIH , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora
8.
Methods Mol Biol ; 462: 307-43, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19160679

RESUMEN

Lipids are actively involved in many cellular processes. Their roles pivot toward determining membrane structure, compartment targeting, and membrane fusion but also regulation of cell signaling via their interactions with proteins and the production of second messengers. As they play a key role in cell signaling, the study of protein-protein interaction and protein conformation change in relationship with their interaction with lipids is of major importance. Until recently, the ability to detect in situ and in real time the dynamics of various biological events and signals without perturbing the cellular environment has been a real challenge. However, the emergence of fluorescence imaging of cells and tissues has allowed the dynamic aspects of the cell to be investigated in a more physiological context than the disassembled model systems employed in traditional biochemical analysis. This chapter highlights some of the many biological applications and uses of frequency- and time-domain fluorescence lifetime imaging microscopy (FLIM) applied to the detection of Förster resonance energy transfer (FRET). The first part describes a FRET system, the second part discusses its study by FLIM, and the third part describes the application of these methods to a panel of biological questions such as (1) spatio-temporal interaction of protein kinase B (PKB) with 3-phosphoinositide dependent protein kinase-1 (PDK1), (2) PKB conformation change, (3) dynamics of PKB activation, (4) interaction of phosphatidylinositol transfer protein (PITP) and phospholipase D (PLD) with lipids.


Asunto(s)
Microscopía Fluorescente/métodos , Fotones , Transducción de Señal , Animales , Anticuerpos Monoclonales/metabolismo , Secuencia de Bases , Línea Celular , Transferencia Resonante de Energía de Fluorescencia , Regulación de la Expresión Génica , Humanos , Metabolismo de los Lípidos , Liposomas/metabolismo , Datos de Secuencia Molecular , Fosfolipasa D/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Fosforilación , Conformación Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Sensibilidad y Especificidad , Coloración y Etiquetado , Factores de Tiempo
9.
Sci Rep ; 9(1): 14527, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601855

RESUMEN

3'-Phosphoinositide-dependent-Kinase-1 (PDK1) is a master regulator whereby its PI3-kinase-dependent dysregulation in human pathologies is well documented. Understanding the direct role for PtdIns(3,4,5)P3 and other anionic phospholipids in the regulation of PDK1 conformational dynamics and its downstream activation remains incomplete. Using advanced quantitative-time-resolved imaging (Fluorescence Lifetime Imaging and Fluorescence Correlation Spectroscopy) and molecular modelling, we show an interplay of antagonistic binding effects of PtdIns(3,4,5)P3 and other anionic phospholipids, regulating activated PDK1 homodimers. We demonstrate that phosphatidylserine maintains PDK1 in an inactive conformation. The dysregulation of the PI3K pathway affects the spatio-temporal and conformational dynamics of PDK1 and the activation of its downstream substrates. We have established a new anionic-phospholipid-dependent model for PDK1 regulation, depicting the conformational dynamics of multiple homodimer states. We show that the dysregulation of the PI3K pathway perturbs equilibrium between the PDK1 homodimer conformations. Our findings provide a role for the PtdSer binding site and its previously unrewarding role in PDK1 downregulation, suggesting a possible therapeutic strategy where the constitutively active dimer conformer of PDK1 may be rendered inactive by small molecules that drive it to its PtdSer-bound conformer.


Asunto(s)
Aniones/química , Fosfolípidos/química , Multimerización de Proteína , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/química , Animales , Proteínas Fluorescentes Verdes/química , Humanos , Lípidos/química , Ratones , Microscopía Confocal , Modelos Moleculares , Células 3T3 NIH , Fosfatidilinositol 3-Quinasas/química , Fosfatos de Fosfatidilinositol , Fosforilación , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/química
10.
Cancer Res ; 66(5): 2834-43, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16510606

RESUMEN

Overexpression and mutation of epidermal growth factor receptors (EGFR) have been shown to be important in the prognosis of several cancers, including head and neck cancers. However, our inability to define the activation status of these and other receptors limits our ability to assess the importance of these pathways and to exploit effectively new molecularly targeted treatments directed at their catalytic activities. Here we describe the use of automated, high-throughput fluorescence lifetime imaging microscopy to measure EGFR autophosphorylation status by fluorescence resonance energy transfer (FRET) in head and neck tumors. We have correlated FRET efficiency with the clinical and survival data. The results from head and neck arrays show that high FRET efficiency is correlated with worsening disease-free survival but not with overall survival. This powerful tool could be exploited as a new independent quantitative prognostic factor in clinical decisions and cancer management.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas/patología , Supervivencia sin Enfermedad , Transferencia Resonante de Energía de Fluorescencia , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunohistoquímica , Estadificación de Neoplasias , Fosforilación , Pronóstico , Análisis de Matrices Tisulares
11.
Biochem J ; 397(1): 109-20, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16669786

RESUMEN

The VHL (von Hippel-Lindau) tumour-suppressor protein forms a multi-protein complex [VCB (pVHL-elongin C-elongin B)-Cul-2 (Cullin-2)] with elongin C, elongin B, Cul-2 and Rbx1, acting as a ubiquitin-ligase (E3) and directing proteasome-dependent degradation of targeted proteins. The alpha-subunit of Hif1alpha (hypoxia-inducible factor 1alpha) is the principal substrate for the VCB-Cul-2 complex; however, other substrates such as aPKC (atypical protein kinase C) have been reported. In the present study, we show with FRET (fluorescence resonance energy transfer) analysis measured by FLIM (fluorescence lifetime imaging microscopy) that PKCdelta and pVHL (VHL protein) interact directly in cells. This occurs through the catalytic domain of PKCdelta (residues 432-508), which appears to interact with two regions of pVHL, residues 113-122 and 130-154. Despite this robust interaction, analysis of the PMA-induced proteasome-dependent degradation of PKCdelta in different RCC (renal cell carcinoma) lines (RCC4, UMRC2 and 786 O) shows that there is no correlation between the degradation of PKCdelta and the presence of active pVHL. Thus, in contrast with aPKC, PKCdelta is not a conventional substrate of the ubiquitin-ligase complex, VCB-Cul-2, and the observed interaction between these two proteins must underlie a distinct signalling output.


Asunto(s)
Proteína Quinasa C-delta/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología , Carcinoma de Células Renales , Catálisis , Dominio Catalítico , Proteínas Cullin/química , Elonguina , Transferencia Resonante de Energía de Fluorescencia , Humanos , Inmunoprecipitación , Neoplasias Renales , Microscopía Fluorescente , Complejos Multiproteicos , Plásmidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Transcripción/química , Células Tumorales Cultivadas , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/química
12.
Sci Rep ; 6: 26142, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27199173

RESUMEN

Strong evidence suggests that phospholipase Cγ1 (PLCγ1) is a suitable target to counteract tumourigenesis and metastasis dissemination. We recently identified a novel signalling pathway required for PLCγ1 activation which involves formation of a protein complex with 3-phosphoinositide-dependent protein kinase 1 (PDK1). In an effort to define novel strategies to inhibit PLCγ1-dependent signals we tested here whether a newly identified and highly specific PDK1 inhibitor, 2-O-benzyl-myo-inositol 1,3,4,5,6-pentakisphosphate (2-O-Bn-InsP5), could affect PDK1/PLCγ1 interaction and impair PLCγ1-dependent cellular functions in cancer cells. Here, we demonstrate that 2-O-Bn-InsP5 interacts specifically with the pleckstrin homology domain of PDK1 and impairs formation of a PDK1/PLCγ1 complex. 2-O-Bn-InsP5 is able to inhibit the epidermal growth factor-induced PLCγ1 phosphorylation and activity, ultimately resulting in impaired cancer cell migration and invasion. Importantly, we report that 2-O-Bn-InsP5 inhibits cancer cell dissemination in zebrafish xenotransplants. This work demonstrates that the PDK1/PLCγ1 complex is a potential therapeutic target to prevent metastasis and it identifies 2-O-Bn-InsP5 as a leading compound for development of anti-metastatic drugs.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Fosfatos de Inositol/farmacología , Fosfolipasa C gamma/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Melanoma/tratamiento farmacológico , Trasplante de Neoplasias , Unión Proteica , Multimerización de Proteína , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Pez Cebra
13.
Cancer Res ; 74(18): 4983-95, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24970478

RESUMEN

Dysregulation of the Akt/PKB pathway has been associated with poor prognosis in several human carcinomas. Current approaches to assess Akt activation rely on intensity-based methods, which are limited by the subjectivity of manual scoring and poor specificity. Here, we report the development of a novel assay using amplified, time-resolved Förster resonance energy transfer (FRET), which is highly specific and sensitive and can be adapted to any protein. Using this approach to analyze primary breast tissue microarrays, we quantified levels of activated pAkt at a spatial resolution that revealed molecular heterogeneity within tumors. High pAkt status assessed by amplified FRET correlated with worse disease-free survival. Our findings support the use of amplified FRET to determine pAkt status in cancer tissues as candidate biomarker for the identification of high-risk patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/enzimología , Transferencia Resonante de Energía de Fluorescencia/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas Proto-Oncogénicas c-akt/análisis , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Humanos , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo
14.
Methods Enzymol ; 506: 225-46, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22341227

RESUMEN

Analytical time resolved Förster resonance energy transfer (FRET) can be exploited for assessing, in cells and tumor micro arrays, the activation status and dynamics of oncoproteins such as epidermal growth factor receptor (EGFR1) and their downstream effectors such as protein kinase B (PKB) and 3-phosphoinositide-dependent protein kinase 1 (PDK1). The outcome of our research involving the application of quantitative imaging for investigating molecular mechanisms of phosphoinositide-dependant enzymes, such as PKB and PDK1, has resulted in a refined model describing the dynamics and regulation of these two oncoproteins in live cells. Our translational research exploits a quantitative FRET method for establishing the activation status of predictive biomarkers in tumor micro arrays. We developed a two-site FRET assay monitored by automated frequency domain Fluorescence lifetime imaging microscopy (FLIM). As a proof of principle, we tested our methodology by assessing EGFR1 activation status in tumor micro arrays from head and neck patients. Our two-site FRET assay, by high-throughput frequency domain FLIM, has great potential to provide prognostic and perhaps predictive biomarkers.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Neoplasias de Cabeza y Cuello/metabolismo , Mapeo de Interacción de Proteínas/métodos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Carcinoma de Células Escamosas/patología , Línea Celular , Neoplasias de Cabeza y Cuello/patología , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Microscopía Fluorescente/métodos , Análisis por Matrices de Proteínas/métodos , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
15.
Nat Cell Biol ; 13(7): 827-37, 2011 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-21642981

RESUMEN

Compartmentalization of signals generated by receptor tyrosine kinase (RTK) endocytosis has emerged as a major determinant of various cell functions. Here, using tumour-associated Met-activating mutations, we demonstrate a direct link between endocytosis and tumorigenicity. Met mutants exhibit increased endocytosis/recycling activity and decreased levels of degradation, leading to accumulation on endosomes, activation of the GTPase Rac1, loss of actin stress fibres and increased levels of cell migration. Blocking endocytosis inhibited mutants' anchorage-independent growth, in vivo tumorigenesis and metastasis while maintaining their activation. One mutant resistant to inhibition by a Met-specific tyrosine kinase inhibitor was sensitive to endocytosis inhibition. Thus, oncogenicity of Met mutants results not only from activation but also from their altered endocytic trafficking, indicating that endosomal signalling may be a crucial mechanism regulating RTK-dependent tumorigenesis.


Asunto(s)
Movimiento Celular , Transformación Celular Neoplásica/metabolismo , Endocitosis , Endosomas/enzimología , Neoplasias Pulmonares/enzimología , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Neoplasias de los Tejidos Blandos/enzimología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Clatrina/metabolismo , Dinaminas/metabolismo , Endocitosis/efectos de los fármacos , Activación Enzimática , Femenino , Proteína Adaptadora GRB2/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Mutación , Células 3T3 NIH , Invasividad Neoplásica , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Interferencia de ARN , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Receptores de Factores de Crecimiento/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/patología , Neoplasias de los Tejidos Blandos/prevención & control , Fibras de Estrés/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
16.
Sci Signal ; 3(145): ra78, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20978239

RESUMEN

3-Phosphoinositide-dependent kinase 1 (PDK1) plays a central role in regulating the activity of protein kinases that are essential for signaling; however, how PDK1 itself is regulated is largely unknown. We found that homodimerization of PDK1 is a spatially and temporally regulated mechanism for controlling PDK1 activity. We used Förster resonance energy transfer monitored by fluorescence lifetime imaging microscopy to observe PDK1 homodimerization in live cells. A pleckstrin homology (PH) domain-dependent, basal dimeric association of PDK1 was increased upon cell stimulation with growth factors; this association was prevented by a phosphatidylinositol 3-kinase inhibitor and by a mutation in, or a complete deletion of, the PH domain of PDK1. The distinct spatial distribution of PDK1 homodimers relative to that of heterodimers of PDK1 and protein kinase B (PKB), and the ability of monomeric mutants of PDK1 to phosphorylate PKB, suggested that the monomer was the active conformation. Mutation of the autophosphorylation residue threonine-513 to glutamate, which was predicted to destabilize the homodimer interface, enhanced the interaction between PDK1 and PKB and the activity of PKB. Through in vitro, time-resolved fluorescence intensity and anisotropy measurements, combined with existing crystal structures and computational molecular modeling, we determined the geometrical arrangement of the PDK1 homodimer. With this approach, we calculated the size of the population of PDK1 dimers in cells. This description of a previously uncharacterized regulatory mechanism for the activation of PDK1 offers possibilities for controlling PDK1 activity therapeutically.


Asunto(s)
Multimerización de Proteína/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Células COS , Chlorocebus aethiops , Activación Enzimática , Transferencia Resonante de Energía de Fluorescencia , Humanos , Ratones , Mutación , Células 3T3 NIH , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína
17.
J Clin Invest ; 120(7): 2516-27, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20530873

RESUMEN

Tumor cell resistance to ionizing radiation and chemotherapy is a major obstacle in cancer therapy. One factor contributing to this is integrin-mediated adhesion to ECM. The adapter protein particularly interesting new cysteine-histidine-rich 1 (PINCH1) is recruited to integrin adhesion sites and promotes cell survival, but the mechanisms underlying this effect are not well understood. Here we have shown that PINCH1 is expressed at elevated levels in human tumors of diverse origins relative to normal tissue. Furthermore, PINCH1 promoted cell survival upon treatment with ionizing radiation in vitro and in vivo by perpetuating Akt1 phosphorylation and activity. Mechanistically, PINCH1 was found to directly bind to protein phosphatase 1alpha (PP1alpha) - an Akt1-regulating protein - and inhibit PP1alpha activity, resulting in increased Akt1 phosphorylation and enhanced radioresistance. Thus, our data suggest that targeting signaling molecules such as PINCH1 that function downstream of focal adhesions (the complexes that mediate tumor cell adhesion to ECM) may overcome radio- and chemoresistance, providing new therapeutic approaches for cancer.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Neoplasias/radioterapia , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales , Adhesión Celular/fisiología , Supervivencia Celular , Cisteína/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Histidina/metabolismo , Humanos , Integrinas/metabolismo , Proteínas con Dominio LIM , Proteínas de la Membrana , Neoplasias/metabolismo , Fosforilación
18.
J Chem Biol ; 2(1): 11-25, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19568789

RESUMEN

New developments regarding the structure and in vivo dynamics of protein kinase B (PKB/Akt) have been recently exposed. Here, we specifically review how the use of multi-disciplinary approaches has resulted in reaching the recent progress made to relate the quaternary structure of PKB to its in vivo function. Using X-ray crystallography, the structure of PKB pleckstrin homology (PH) and kinase domains was determined separately. The molecular mechanisms involved in (a) the binding of the phosphoinositides to the PH domain and (b) the activation of the kinase with the rearrangement of the catalytic site and substrate binding were determined. In vitro, nuclear magnetic resonance and circular dychroism studies gave complementary information on the interaction of the PH domain with the phosphoinositides. However, the molecular nature and the function of the interactions between the PKB domains could not be deduced from the X-ray data since the full-length PKB has not been crystallised. In vitro, dynamic information on the inter-domain conformational changes related to PKB activation states emerged with the use of tandem mass spectrometry. Cell imaging and Förster resonance energy transfer provided in vivo dynamics. Molecular modelling and dynamic simulations in conjunction with mutagenesis and biochemical analysis were used to investigate the complex interactions between the PKB domains in vivo and understand at the molecular level how it linked to its activity. The compilation of the information obtained on the 3-D structure and the spatiotemporal dynamics of this widely studied oncogene could be applied to the study of other proteins. This inter-disciplinary approach led to a more profound understanding of PKB complex activation mechanism in vivo that will shed light onto new ideas and possibilities for modulating its activity.

19.
PLoS One ; 3(8): e2881, 2008 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-18682844

RESUMEN

BACKGROUND: The response rate to EGFR tyrosine kinase inhibitors (TKIs) may be poor and unpredictable in cancer patients with EGFR expression itself being an inadequate response indicator. There is limited understanding of the mechanisms underlying this resistance. Furthermore, although TKIs suppress the growth of HER2-overexpressing breast tumor cells, they do not fully inhibit HER2 oncogenic function at physiological doses. METHODOLOGY AND PRINCIPAL FINDINGS: Here we have provided a molecular mechanism of how HER2 oncogenic function escapes TKIs' inhibition via alternative HER receptor activation as a result of autocrine ligand release. Using both Förster Resonance Energy Transfer (FRET) which monitors in situ HER receptor phosphorylation as well as classical biochemical analysis, we have shown that the specific tyrosine kinase inhibitors (TKIs) of EGFR, AG1478 and Iressa (Gefitinib) decreased EGFR and HER3 phosphorylation through the inhibition of EGFR/HER3 dimerization. Consequent to this, we demonstrate that cleavage of HER4 and dimerization of HER4/HER2 occur together with reactivation of HER3 via HER2/HER3, leading to persistent HER2 phosphorylation in the now resistant, surviving cells. These drug treatment-induced processes were found to be mediated by the release of ligands including heregulin and betacellulin that activate HER3 and HER4 via HER2. Whereas an anti-betacellulin antibody in combination with Iressa increased the anti-proliferative effect in resistant cells, ligands such as heregulin and betacellulin rendered sensitive SKBR3 cells resistant to Iressa. CONCLUSIONS AND SIGNIFICANCE: These results demonstrate the role of drug-induced autocrine events leading to the activation of alternative HER receptors in maintaining HER2 phosphorylation and in mediating resistance to EGFR tyrosine kinase inhibitors (TKIs) in breast cancer cells, and hence specify treatment opportunities to overcome resistance in patients.


Asunto(s)
Neoplasias de la Mama/enzimología , Receptores ErbB/metabolismo , Genes erbB-2/fisiología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Tirfostinos/farmacología , Betacelulina , Neoplasias de la Mama/genética , Línea Celular Tumoral , Activación Enzimática , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neurregulina-1/metabolismo , Fosforilación , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Quinazolinas , Receptor ErbB-4
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA